551
|
Qu J, Mei Q, Chen L, Zhou J. Chimeric antigen receptor (CAR)-T-cell therapy in non-small-cell lung cancer (NSCLC): current status and future perspectives. Cancer Immunol Immunother 2021; 70:619-631. [PMID: 33025047 PMCID: PMC7907037 DOI: 10.1007/s00262-020-02735-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022]
Abstract
There has been a rapid progress in developing genetically engineered T cells in recent years both in basic and clinical cancer studies. Chimeric antigen receptor (CAR)-T cells exert an immune response against various cancers, including the non-small-cell lung cancer (NSCLC). As novel agents of immunotherapy, CAR-T cells show great promise for NSCLC. However, targeting specific antigens in NSCLC with engineered CAR-T cells is complicated because of a lack of tumor-specific antigens, the immunosuppressive tumor microenvironment, low levels of infiltration of CAR-T cells into tumor tissue, and tumor antigen escape. Meanwhile, the clinical application of CAR-T cells remains limited due to the cases of on-target/off-tumor and neurological toxicity, as well as cytokine release syndrome. Hence, optimal CAR-T-cell design against NSCLC is urgently needed. In this review, we describe the basic structure and generation of CAR-T cells and summarize the common tumor-associated antigens targeted in clinical trials on CAR-T-cell therapy for NSCLC, as well as point out current challenges and novel strategies. Although many obstacles remain, the new/next generation of CARs show much promise. Taken together, research on CAR-T cells for the treatment of NSCLC is underway and has yielded promising preliminary results both in basic and pre-clinical medicine. More pre-clinical experiments and clinical trials are, therefore, warranted.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Biomarkers, Tumor
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Clinical Trials as Topic
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Lung Neoplasms/diagnosis
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Prognosis
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Treatment Outcome
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
- Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Quanhui Mei
- Department of Intensive Care Unit, The First People's Hospital of Changde City, Changde, Hunan, 415003, People's Republic of China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China.
- Stowers Institute for Medical Research, 1000 E 50th Street, Kansas City, MO, 64110, USA.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Centre, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China.
| |
Collapse
|
552
|
IL-2 regulates tumor-reactive CD8 + T cell exhaustion by activating the aryl hydrocarbon receptor. Nat Immunol 2021; 22:358-369. [PMID: 33432230 DOI: 10.1038/s41590-020-00850-9] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/26/2020] [Indexed: 01/29/2023]
Abstract
CD8+ T cell exhaustion dampens antitumor immunity. Although several transcription factors have been identified that regulate T cell exhaustion, the molecular mechanisms by which CD8+ T cells are triggered to enter an exhausted state remain unclear. Here, we show that interleukin-2 (IL-2) acts as an environmental cue to induce CD8+ T cell exhaustion within tumor microenvironments. We find that a continuously high level of IL-2 leads to the persistent activation of STAT5 in CD8+ T cells, which in turn induces strong expression of tryptophan hydroxylase 1, thus catalyzing the conversion to tryptophan to 5-hydroxytryptophan (5-HTP). 5-HTP subsequently activates AhR nuclear translocation, causing a coordinated upregulation of inhibitory receptors and downregulation of cytokine and effector-molecule production, thereby rendering T cells dysfunctional in the tumor microenvironment. This molecular pathway is not only present in mouse tumor models but is also observed in people with cancer, identifying IL-2 as a novel inducer of T cell exhaustion.
Collapse
|
553
|
Schoenfeld AJ, O'Cearbhaill RE. How Do We Meet the Challenge of Chimeric Antigen Receptor T-Cell Therapy for Solid Tumors? Cancer J 2021; 27:134-142. [PMID: 33750073 PMCID: PMC8457037 DOI: 10.1097/ppo.0000000000000516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT Immune checkpoint inhibition has vastly improved the treatment of solid tumors, but most patients do not experience durable clinical benefit, so novel immunotherapeutic approaches are needed. Autologous T cells genetically engineered to express chimeric antigen receptors (CARs) have led to unprecedented clinical success in hematologic malignancies, and increasing efforts are actively being pursued to translate these benefits to the solid tumor arena. However, solid tumors present unique challenges for CAR T-cell development. In this review, we examine the potential barriers to progress and present emerging approaches to overcome these challenges with CAR therapy in solid tumors.
Collapse
Affiliation(s)
- Adam J Schoenfeld
- From the Cellular Therapy Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Department of Medicine, Weill Cornell Medical College, New York, NY
| | | |
Collapse
|
554
|
Brayshaw LL, Martinez-Fleites C, Athanasopoulos T, Southgate T, Jespers L, Herring C. The role of small molecules in cell and gene therapy. RSC Med Chem 2021; 12:330-352. [PMID: 34046619 PMCID: PMC8130622 DOI: 10.1039/d0md00221f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/25/2020] [Indexed: 01/22/2023] Open
Abstract
Cell and gene therapies have achieved impressive results in the treatment of rare genetic diseases using gene corrected stem cells and haematological cancers using chimeric antigen receptor T cells. However, these two fields face significant challenges such as demonstrating long-term efficacy and safety, and achieving cost-effective, scalable manufacturing processes. The use of small molecules is a key approach to overcome these barriers and can benefit cell and gene therapies at multiple stages of their lifecycle. For example, small molecules can be used to optimise viral vector production during manufacturing or used in the clinic to enhance the resistance of T cell therapies to the immunosuppressive tumour microenvironment. Here, we review current uses of small molecules in cell and gene therapy and highlight opportunities for medicinal chemists to further consolidate the success of cell and gene therapies.
Collapse
Affiliation(s)
- Lewis L Brayshaw
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Carlos Martinez-Fleites
- Protein Degradation Group, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Takis Athanasopoulos
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Thomas Southgate
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Laurent Jespers
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Christopher Herring
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| |
Collapse
|
555
|
Pre-conditioning modifies the TME to enhance solid tumor CAR T cell efficacy and endogenous protective immunity. Mol Ther 2021; 29:2335-2349. [PMID: 33647456 PMCID: PMC8261088 DOI: 10.1016/j.ymthe.2021.02.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/23/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has led to impressive clinical responses in patients with hematological malignancies; however, its effectiveness in patients with solid tumors has been limited. While CAR T cells for the treatment of advanced prostate and pancreas cancer, including those targeting prostate stem cell antigen (PSCA), are being clinically evaluated and are anticipated to show bioactivity, their safety and the impact of the immunosuppressive tumor microenvironment (TME) have not been faithfully explored preclinically. Using a novel human PSCA knockin (hPSCA-KI) immunocompetent mouse model, we evaluated the safety and therapeutic efficacy of PSCA-CAR T cells. We demonstrated that cyclophosphamide (Cy) pre-conditioning significantly modified the immunosuppressive TME and was required to uncover the efficacy of PSCA-CAR T cells in metastatic prostate and pancreas cancer models, with no observed toxicities in normal tissues with endogenous expression of PSCA. This combination dampened the immunosuppressive TME, generated pro-inflammatory myeloid and T cell signatures in tumors, and enhanced the recruitment of antigen-presenting cells, as well as endogenous and adoptively transferred T cells, resulting in long-term anti-tumor immunity.
Collapse
|
556
|
Kovalovsky D, Yoon JH, Cyr MG, Simon S, Voynova E, Rader C, Wiestner A, Alejo J, Pittaluga S, Gress RE. Siglec-6 is a target for chimeric antigen receptor T-cell treatment of chronic lymphocytic leukemia. Leukemia 2021; 35:2581-2591. [PMID: 33633313 PMCID: PMC8384967 DOI: 10.1038/s41375-021-01188-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 01/02/2023]
Abstract
The only current curative treatment for chronic lymphocytic leukemia (CLL) is allogenic hematopoietic stem cell transplantation. Chimeric antigen receptor treatment targeting CD19 for CLL achieved some complete responses, suggesting the need for alternative or combinational therapies to achieve a more robust response. In this work, we evaluated CAR-T cells specific for Siglec-6, an antigen expressed in CLL, as a novel CAR-T cell treatment for CLL. We found that detection of SIGLEC6 mRNA and Siglec-6 protein is highly restricted to placenta and immune cells in other tissues and it is not expressed in hematopoietic stem cells. We generated CAR-T cells specific for Siglec-6 based on the sequence of the fully human anti-Siglec-6 antibody (JML1), which was identified in a CLL patient that was cured after allo-hematopoietic stem cell transplantation (alloHSCT), and observed that it specifically targeted CLL cells in vitro and in a xenograft mouse model. Interestingly, a short hinge region increased the activity of CAR-T cells to target cells expressing higher Siglec-6 levels but similarly targeted CLL cells expressing lower Siglec-6 levels in vitro and in vivo. Our results identify a novel CAR-T cell therapy for CLL and establish Siglec-6 as a possible target for immunotherapy.
Collapse
Affiliation(s)
- Damian Kovalovsky
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA.
| | - Jeong Heon Yoon
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Matthew G Cyr
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Samantha Simon
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Elisaveta Voynova
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Adrian Wiestner
- Laboratory of Lymphoid Malignancies, National Heart, Lung and Blood Institute. NIH, Bethesda, MD, 20892, USA
| | - Julie Alejo
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Ronald E Gress
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
557
|
Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, Hegde M, Ruella M, Savoldo B, Shah NN, Turtle CJ, Wayne AS, Abou-El-Enein M. Immunogenicity of CAR T cells in cancer therapy. Nat Rev Clin Oncol 2021; 18:379-393. [PMID: 33633361 PMCID: PMC8923136 DOI: 10.1038/s41571-021-00476-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Patient-derived T cells genetically reprogrammed to express CD19-specific chimeric antigen receptors (CARs) have shown remarkable clinical responses and are commercially available for the treatment of patients with certain advanced-stage B cell malignancies. Nonetheless, several trials have revealed pre-existing and/or treatment-induced immune responses to the mouse-derived single-chain variable fragments included in these constructs. These responses might have contributed to both treatment failure and the limited success of redosing strategies observed in some patients. Data from early phase clinical trials suggest that CAR T cells are also associated with immunogenicity-related events in patients with solid tumours. Generally, the clinical implications of anti-CAR immune responses are poorly understood and highly variable between different CAR constructs and malignancies. These observations highlight an urgent need to uncover the mechanisms of immunogenicity in patients receiving CAR T cells and develop validated assays to enable clinical detection. In this Review, we describe the current clinical evidence of anti-CAR immune responses and discuss how new CAR T cell technologies might impact the risk of immunogenicity. We then suggest ways to reduce the risks of anti-CAR immune responses to CAR T cell products that are advancing towards the clinic. Finally, we summarize measures that investigators could consider in order to systematically monitor and better comprehend the possible effects of immunogenicity during trials involving CAR T cells as well as in routine clinical practice.
Collapse
Affiliation(s)
- Dimitrios L Wagner
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Enrico Fritsche
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael A Pulsipher
- Section of Transplantation and Cellular Therapy, Children's Hospital Los Angeles Cancer and Blood Disease Institute, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Nabil Ahmed
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Mohamad Hamieh
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, NY, USA
| | - Meenakshi Hegde
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania Philadelphia, Philadelphia, PA, USA.,Division of Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alan S Wayne
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mohamed Abou-El-Enein
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Division of Medical Oncology, Department of Medicine, and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. .,Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
558
|
Fei Z, Fan Q, Dai H, Zhou X, Xu J, Ma Q, Maruyama A, Wang C. Physiologically triggered injectable red blood cell-based gel for tumor photoablation and enhanced cancer immunotherapy. Biomaterials 2021; 271:120724. [PMID: 33636549 DOI: 10.1016/j.biomaterials.2021.120724] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/01/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
Hydrogels are widely used for drug delivery and tissue engineering. Here we developed a simple injectable red blood cells (RBCs)-based gel for cancer photo-immunotherapy. We find that subcutaneous injected homologous RBCs could form hydrogel-like composition in mice, due to the infiltrated platelets and thrombin under physiological environment. In addition, the formed RBC-gel has photothermal effect under NIR laser exposure on account of deep reddish color. In mice bearing CT26 tumors, we demonstrate photo-immunotherapy of cancer by local injection of imiquimod (R837) adjuvant engineered RBCs. The photothermal effect of the in situ formed RBC-gel effectively burns tumor to release tumor-associated antigens (TAAs), promotes the release of R837 from RBCs to the tumor draining lymph node, thereby activating the lymph node-resident antigen-presenting cells (APCs) remarkably. A durable systemic immune response is induced following the combination treatment of the primary tumor. 100% mice rejected tumor rechallenge and are survived at least 250 days without any detectable tumors. Our strategy highlights the RBCs, the most common type of cell in our blood, as the hydrogel for drug delivery and cancer photo-immunotherapy.
Collapse
Affiliation(s)
- Ziying Fei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qin Fan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China; Key Laboratory for Organic Electronics & Information Displays (KLOEID), Institute of Advanced Materials (IAM) and School of Materials Science and Engineering, Nanjing University of Posts & Telecommunications, Nanjing, 210000, China.
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuanfang Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Atsushi Maruyama
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259 B-57, Nagatsuta, Yokohama, 226-8501, Japan.
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
559
|
Tan X, Letendre JH, Collins JJ, Wong WW. Synthetic biology in the clinic: engineering vaccines, diagnostics, and therapeutics. Cell 2021; 184:881-898. [PMID: 33571426 PMCID: PMC7897318 DOI: 10.1016/j.cell.2021.01.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/17/2022]
Abstract
Synthetic biology is a design-driven discipline centered on engineering novel biological functions through the discovery, characterization, and repurposing of molecular parts. Several synthetic biological solutions to critical biomedical problems are on the verge of widespread adoption and demonstrate the burgeoning maturation of the field. Here, we highlight applications of synthetic biology in vaccine development, molecular diagnostics, and cell-based therapeutics, emphasizing technologies approved for clinical use or in active clinical trials. We conclude by drawing attention to recent innovations in synthetic biology that are likely to have a significant impact on future applications in biomedicine.
Collapse
Affiliation(s)
- Xiao Tan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA; Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA
| | - Justin H Letendre
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - James J Collins
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Synthetic Biology Center, MIT, 77 Massachusetts Ave., Cambridge, MA 02139, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
560
|
Goto S, Sakoda Y, Adachi K, Sekido Y, Yano S, Eto M, Tamada K. Enhanced anti-tumor efficacy of IL-7/CCL19-producing human CAR-T cells in orthotopic and patient-derived xenograft tumor models. Cancer Immunol Immunother 2021; 70:2503-2515. [PMID: 33559069 DOI: 10.1007/s00262-021-02853-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/05/2021] [Indexed: 01/03/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has impressive efficacy in hematological malignancies, but its application in solid tumors remains a challenge. Multiple hurdles associated with the biological and immunological features of solid tumors currently limit the application of CAR-T cells in the treatment of solid tumors. Using syngeneic mouse models, we recently reported that CAR-T cells engineered to concomitantly produce interleukin (IL)-7 and chemokine (C-C motif) ligand 19 (CCL19)-induced potent anti-tumor efficacy against solid tumors through an improved ability of migration and proliferation even in an immunosuppressive tumor microenvironment. In this study, for a preclinical evaluation preceding clinical application, we further explored the potential of IL-7/CCL19-producing human CAR-T cells using models that mimic the clinical features of solid tumors. Human anti-mesothelin CAR-T cells producing human IL-7/CCL19 achieved complete eradication of orthotopic pre-established malignant mesothelioma and prevented a relapse of tumors with downregulated antigen expression. Moreover, mice with patient-derived xenograft of mesothelin-positive pancreatic cancers exhibited significant inhibition of tumor growth and prolonged survival following treatment with IL-7/CCL19-producing CAR-T cells, compared to treatment with conventional CAR-T cells. Transfer of IL-7/CCL19-producing CAR-T cells resulted in an increase in not only CAR-T cells but also non-CAR-T cells within the tumor tissues and downregulated the expression of exhaustion markers, including PD-1 and TIGIT, on the T cells. Taken together, our current study elucidated the exceptional anti-tumor efficacy of IL-7/CCL19-producing human CAR-T cells and their potential for clinical application in the treatment of patients with solid tumors.
Collapse
Affiliation(s)
- Shunsuke Goto
- Department of Immunology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.,Department of Urology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Yukimi Sakoda
- Department of Immunology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Keishi Adachi
- Department of Immunology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshitaka Sekido
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
561
|
Leick MB, Maus MV, Frigault MJ. Clinical Perspective: Treatment of Aggressive B Cell Lymphomas with FDA-Approved CAR-T Cell Therapies. Mol Ther 2021; 29:433-441. [PMID: 33130313 PMCID: PMC7854294 DOI: 10.1016/j.ymthe.2020.10.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/10/2020] [Accepted: 10/24/2020] [Indexed: 02/08/2023] Open
Abstract
Large B cell lymphoma (LBCL) is curable with standard chemo-immunotherapy in the majority of cases. However, patients with primary refractory or relapsed disease have historically had limited treatment options. Two gene-modified chimeric antigen receptor (CAR)-T cell therapies have now been approved for these indications. The clinical decisions and management surrounding these gene-modified "living drugs" are nuanced and complex. In this article, we discuss the evolving evidence supporting the use of these CAR-T cells, including patient selection, screening procedures, special populations, bridging therapy, lymphodepletion, clinical management, relapse, and follow up.
Collapse
Affiliation(s)
- Mark B Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA, USA.
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
562
|
T-Cell Dysfunction as a Limitation of Adoptive Immunotherapy: Current Concepts and Mitigation Strategies. Cancers (Basel) 2021; 13:cancers13040598. [PMID: 33546277 PMCID: PMC7913380 DOI: 10.3390/cancers13040598] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary T cells are immune cells that can be used to target infections or cancers. Adoptive T-cell immunotherapy leverages these properties and/or confers new features to T cells through ex vivo manipulations prior to their use in patients. However, as a “living drug,” the function of these cells can be hampered by several built-in physiological constraints and external factors that limit their efficacy. Manipulating T cells ex vivo can impart dysfunctional features to T cells through repeated stimulations and expansion, but it also offers many opportunities to improve the therapeutic potential of these cells, including emerging interventions to prevent or reverse T-cell dysfunction developing ex vivo or after transfer in patients. This review outlines the various forms of T-cell dysfunction, emphasizes how it affects various types of T-cell immunotherapy approaches, and describes current and anticipated strategies to limit T-cell dysfunction. Abstract Over the last decades, cellular immunotherapy has revealed its curative potential. However, inherent physiological characteristics of immune cells can limit the potency of this approach. Best defined in T cells, dysfunction associated with terminal differentiation, exhaustion, senescence, and activation-induced cell death, undermine adoptive cell therapies. In this review, we concentrate on how the multiple mechanisms that articulate the various forms of immune dysfunction impact cellular therapies primarily involving conventional T cells, but also other lymphoid subtypes. The repercussions of immune cell dysfunction across the full life cycle of cell therapy, from the source material, during manufacturing, and after adoptive transfer, are discussed, with an emphasis on strategies used during ex vivo manipulations to limit T-cell dysfunction. Applicable to cellular products prepared from native and unmodified immune cells, as well as genetically engineered therapeutics, the understanding and potential modulation of dysfunctional features are key to the development of improved cellular immunotherapies.
Collapse
|
563
|
Jiang L, Jung S, Zhao J, Kasinath V, Ichimura T, Joseph J, Fiorina P, Liss AS, Shah K, Annabi N, Joshi N, Akama TO, Bromberg JS, Kobayashi M, Uchimura K, Abdi R. Simultaneous targeting of primary tumor, draining lymph node, and distant metastases through high endothelial venule-targeted delivery. NANO TODAY 2021; 36:101045. [PMID: 33391389 PMCID: PMC7774643 DOI: 10.1016/j.nantod.2020.101045] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cancer patients with malignant involvement of tumor-draining lymph nodes (TDLNs) and distant metastases have the poorest prognosis. A drug delivery platform that targets the primary tumor, TDLNs, and metastatic niches simultaneously, remains to be developed. Here, we generated a novel monoclonal antibody (MHA112) against peripheral node addressin (PNAd), a family of glycoproteins expressed on high endothelial venules (HEVs), which are present constitutively in the lymph nodes (LNs) and formed ectopically in the tumor stroma. MHA112 was endocytosed by PNAd-expressing cells, where it passed through the lysosomes. MHA112 conjugated antineoplastic drug Paclitaxel (Taxol) (MHA112-Taxol) delivered Taxol effectively to the HEV-containing tumors, TDLNs, and metastatic lesions. MHA112-Taxol treatment significantly reduced primary tumor size as well as metastatic lesions in a number of mouse and human tumor xenografts tested. These data, for the first time, indicate that human metastatic lesions contain HEVs and provide a platform that permits simultaneous targeted delivery of antineoplastic drugs to the three key sites of primary tumor, TDLNs, and metastases.
Collapse
Affiliation(s)
- Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sungwook Jung
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Fiorina
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew S. Liss
- Department of Surgery and the Andrew L. Warshaw, MD Institute for Pancreatic Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard medical School, Boston, MA, 02115, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoya O. Akama
- Department of Pharmacology, Kansai Medical University, Osaka, 570-8506, Japan
| | - Jonathan S. Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Kenji Uchimura
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- CNRS, UMR 8576, Unit of Glycobiology Structures and Functions, University of Lille, F-59000 Lille, France
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
564
|
Crotty EE, Downey KM, Ferrerosa LM, Flores CT, Hegde B, Raskin S, Hwang EI, Vitanza NA, Okada H. Considerations when treating high-grade pediatric glioma patients with immunotherapy. Expert Rev Neurother 2021; 21:205-219. [PMID: 33225764 PMCID: PMC7880880 DOI: 10.1080/14737175.2020.1855144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Children with high-grade gliomas (pHGGs) represent a clinical population in substantial need of new therapeutic options given the inefficacy and toxicity of current standard-of-care modalities. Although immunotherapy has emerged as a promising modality, it has yet to elicit a significant survival benefit for pHGG patients. While preclinical studies address a variety of underlying challenges, translational clinical trial design and management also need to reflect the most updated progress and lessons from the field. AREAS COVERED The authors will focus our discussion on the design of clinical trials, the management of potential toxicities, immune monitoring, and novel biomarkers. Clinical trial design should integrate appropriate patient populations, novel, and preclinically optimized trial design, and logical treatment combinations, particularly those which synergize with standard of care modalities. However, there are caveats due to the nature of immunotherapy trials, such as patient selection bias, evidenced by the frequent exclusion of patients on high-dose corticosteroids. Robust immune-modulating effects of modern immunotherapy can have toxicities. As such, it is important to understand and manage these, especially in pHGG patients. EXPERT OPINION Adequate integration of these considerations should allow us to effectively gain insights on biological activity, safety, and biomarkers associated with benefits for patients.
Collapse
Affiliation(s)
- Erin E. Crotty
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Kira M. Downey
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Lauren M. Ferrerosa
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCSF Benioff Children’s Hospital, Oakland, 747 52nd Street, Oakland, CA, USA
| | | | - Bindu Hegde
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Scott Raskin
- Children’s National Hospital, Washington, DC, USA
| | | | - Nicholas A. Vitanza
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Hideho Okada
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Research Center, University of California San Francisco, San Francisco, CA, USA
- The Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
- Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
565
|
Sánchez‐Martínez D, Gutiérrez‐Agüera F, Romecin P, Vinyoles M, Palomo M, Tirado N, Zanetti SR, Juan M, Carlet M, Jeremias I, Menéndez P. Enforced sialyl-Lewis-X (sLeX) display in E-selectin ligands by exofucosylation is dispensable for CD19-CAR T-cell activity and bone marrow homing. Clin Transl Med 2021; 11:e280. [PMID: 33634970 PMCID: PMC7901721 DOI: 10.1002/ctm2.280] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022] Open
Abstract
CD19-directed chimeric antigen receptors (CAR) T cells induce impressive rates of complete response in advanced B-cell malignancies, specially in B-cell acute lymphoblastic leukemia (B-ALL). However, CAR T-cell-treated patients eventually progress due to poor CAR T-cell persistence and/or disease relapse. The bone marrow (BM) is the primary location for acute leukemia. The rapid/efficient colonization of the BM by systemically infused CD19-CAR T cells might enhance CAR T-cell activity and persistence, thus, offering clinical benefits. Circulating cells traffic to BM upon binding of tetrasaccharide sialyl-Lewis X (sLeX)-decorated E-selectin ligands (sialofucosylated) to the E-selectin receptor expressed in the vascular endothelium. sLeX-installation in E-selectin ligands is achieved through an ex vivo fucosylation reaction. Here, we sought to characterize the basal and cell-autonomous display of sLeX in CAR T-cells activated using different cytokines, and to assess whether exofucosylation of E-selectin ligands improves CD19-CAR T-cell activity and BM homing. We report that cell-autonomous sialofucosylation (sLeX display) steadily increases in culture- and in vivo-expanded CAR T cells, and that, the cytokines used during T-cell activation influence both the degree of such endogenous sialofucosylation and the CD19-CAR T-cell efficacy and persistence in vivo. However, glycoengineered enforced sialofucosylation of E-selectin ligands was dispensable for CD19-CAR T-cell activity and BM homing in multiple xenograft models regardless the cytokines employed for T-cell expansion, thus, representing a dispensable strategy for CD19-CAR T-cell therapy.
Collapse
Affiliation(s)
- Diego Sánchez‐Martínez
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Francisco Gutiérrez‐Agüera
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Paola Romecin
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Meritxell Vinyoles
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Marta Palomo
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Néstor Tirado
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Samanta Romina Zanetti
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Manel Juan
- Servei d'ImmunologiaHospital Clínic de BarcelonaBarcelonaSpain
| | - Michela Carlet
- Department of Apoptosis in Hematopoietic Stem Cells, Helmholtz Center MunichGerman Center for Environmental Health (HMGU)MunichGermany
- Department of PediatricsDr von Hauner Children's Hospital, LMUMunichGermany
| | - Irmela Jeremias
- Department of Apoptosis in Hematopoietic Stem Cells, Helmholtz Center MunichGerman Center for Environmental Health (HMGU)MunichGermany
- Department of PediatricsDr von Hauner Children's Hospital, LMUMunichGermany
| | - Pablo Menéndez
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of MedicineUniversity of BarcelonaBarcelonaSpain
- Department of Biomedicine, School of MedicineUniversity of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red‐Oncología (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| |
Collapse
|
566
|
Lü P, Qiu S, Pan Y, Yu F, Chen K. Preclinical Chimeric Antibody Chimeric Antigen Receptor T Cell Progress in Digestive System Cancers. Cancer Biother Radiopharm 2021; 36:307-315. [PMID: 33481647 DOI: 10.1089/cbr.2020.4089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Digestive system cancers, including hepatocellular carcinoma, colorectal and gastric tumors, are characterized by high rates of incidence and mortality. Digestive cancers are difficult to diagnose during the early stages, and the side effects of chemotherapy are often severe and may outweigh the therapeutic benefits. Chimeric antibody chimeric antigen receptor T cell (CAR-T) therapy, a novel immunotherapy, has achieved excellent results for the treatment of hematological tumors. However, CAR-T treatment of solid tumors has struggled due to a lack of target specificity, a difficult tumor microenvironment, and T cell homing. Despite the challenges, CAR-T treatment of digestive cancers is progressing. Combining CAR-T with other targets and/or modifying the CAR may represent the most promising approaches for future treatment of digestive cancers.
Collapse
Affiliation(s)
- Peng Lü
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China.,School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, China
| | - Songlin Qiu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ye Pan
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
567
|
Shu R, Evtimov VJ, Hammett MV, Nguyen NYN, Zhuang J, Hudson PJ, Howard MC, Pupovac A, Trounson AO, Boyd RL. Engineered CAR-T cells targeting TAG-72 and CD47 in ovarian cancer. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:325-341. [PMID: 33614914 PMCID: PMC7868933 DOI: 10.1016/j.omto.2021.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/09/2021] [Indexed: 02/04/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have revolutionized blood cancer immunotherapy; however, their efficacy against solid tumors has been limited. A common mechanism of tumor escape from single target therapies is downregulation or mutational loss of the nominal epitope. Targeting multiple antigens may thus improve the effectiveness of CAR immunotherapies. We generated dual CAR-T cells targeting two tumor antigens: TAG-72 (tumor-associated glycoprotein 72) and CD47. TAG-72 is a pan-adenocarcinoma oncofetal antigen, highly expressed in ovarian cancers, with increased expression linked to disease progression. CD47 is ubiquitously overexpressed in multiple tumor types, including ovarian cancer; it is a macrophage “don’t eat me” signal. However, CD47 is also expressed on many normal cells. To avoid this component of the dual CAR-T cells killing healthy tissue, we designed a truncated CD47 CAR devoid of intracellular signaling domains. The CD47 CAR facilitates binding to CD47+ cells, increasing the prospect of TAG-72+ cell elimination via the TAG-72 CAR. Furthermore, we could reduce the damage to normal tissue by monomerizing the CD47 CAR. Our results indicate that the co-expression of the TAG-72 CAR and the CD47-truncated monomer CAR on T cells could be an effective, dual CAR-T cell strategy for ovarian cancer, also applicable to other adenocarcinomas.
Collapse
Affiliation(s)
- Runzhe Shu
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia
| | | | | | | | - Junli Zhuang
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia
| | - Peter J Hudson
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia.,Avipep Pty, Ltd., Parkville, VIC 3052, Australia
| | | | | | - Alan O Trounson
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | | |
Collapse
|
568
|
CXCR5 CAR-T cells simultaneously target B cell non-Hodgkin's lymphoma and tumor-supportive follicular T helper cells. Nat Commun 2021; 12:240. [PMID: 33431832 PMCID: PMC7801647 DOI: 10.1038/s41467-020-20488-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/01/2020] [Indexed: 12/19/2022] Open
Abstract
CAR-T cell therapy targeting CD19 demonstrated strong activity against advanced B cell leukemia, however shows less efficacy against lymphoma with nodal dissemination. To target both B cell Non-Hodgkin’s lymphoma (B-NHLs) and follicular T helper (Tfh) cells in the tumor microenvironment (TME), we apply here a chimeric antigen receptor (CAR) that recognizes human CXCR5 with high avidity. CXCR5, physiologically expressed on mature B and Tfh cells, is also highly expressed on nodal B-NHLs. Anti-CXCR5 CAR-T cells eradicate B-NHL cells and lymphoma-supportive Tfh cells more potently than CD19 CAR-T cells in vitro, and they efficiently inhibit lymphoma growth in a murine xenograft model. Administration of anti-murine CXCR5 CAR-T cells in syngeneic mice specifically depletes endogenous and malignant B and Tfh cells without unexpected on-target/off-tumor effects. Collectively, anti-CXCR5 CAR-T cells provide a promising treatment strategy for nodal B-NHLs through the simultaneous elimination of lymphoma B cells and Tfh cells of the tumor-supporting TME. CAR-T cell therapy targeting CD19 is not as efficient to treat lymphoma with nodal dissemination as it is for B cell leukaemia. Here, the authors generate CAR-T cells against CXCR5 and show they inhibit tumour growth by depleting both B and follicular T helper cells in lymphoma models.
Collapse
|
569
|
Riggan L, Shah S, O’Sullivan TE. Arrested development: suppression of NK cell function in the tumor microenvironment. Clin Transl Immunology 2021; 10:e1238. [PMID: 33456775 PMCID: PMC7797224 DOI: 10.1002/cti2.1238] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are cytotoxic innate lymphocytes that protect against viral infection and tumor metastasis. Despite their inherent ability to kill a broad range of virally infected, stressed and transformed cells, low numbers of dysfunctional NK cells are often observed in many advanced solid human cancers. Here, we review the potential mechanisms that influence suboptimal mature NK cell recruitment and function in the tumor microenvironment (TME) of solid tumors. We further highlight current immunotherapy approaches aimed to circumvent NK cell dysfunction and discuss next-generation strategies to enhance adoptive NK cell therapy through targeting intrinsic and extrinsic checkpoints the regulate NK cell functionality in the TME. Understanding the mechanisms that drive NK cell dysfunction in the TME will lead to novel immunotherapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Luke Riggan
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Molecular Biology InstituteUniversity of CaliforniaLos AngelesCAUSA
| | - Siya Shah
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Timothy E O’Sullivan
- Department of Microbiology, Immunology, and Molecular GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Molecular Biology InstituteUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
570
|
Warda W, Da Rocha MN, Trad R, Haderbache R, Salma Y, Bouquet L, Roussel X, Nicod C, Deschamps M, Ferrand C. Overcoming target epitope masking resistance that can occur on low-antigen-expresser AML blasts after IL-1RAP chimeric antigen receptor T cell therapy using the inducible caspase 9 suicide gene safety switch. Cancer Gene Ther 2021; 28:1365-1375. [PMID: 33414517 PMCID: PMC8636256 DOI: 10.1038/s41417-020-00284-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/27/2020] [Accepted: 12/04/2020] [Indexed: 12/31/2022]
Abstract
Although chimeric antigen receptor CAR) T cell immunotherapies are an undeniable and unequivocal success, knowledge obtained from the monitoring of the first clinical trials targeting the CD19 antigen in B malignancies, either refractory/relapsed acute lymphoid leukemia (ALL) or lymphomas, contributed to the identification of tumor cell escape in about 30–50% of B-ALL. Resistance occurred due to loss of surface expression of the antigen (rCD19−) or to the early disappearance or inactivation of CAR T cells (rCD19+). In a recently reported clinical case, rCD19− relapse resulted from masking of the antigen by the CAR at the surface of B-ALL leukemia cells following the unexpected viral transduction of a leukemic cell present in the cytapheresis sample. The objective of this work was to reproduce this epitope-masking resistance model, in the context of acute myeloid leukemia (AML), based on our immunotherapeutic CAR T cell model targeting the accessory protein of the interleukin-1 receptor (IL-1RAP) expressed by leukemic stem cells. As AML primary blasts express different levels of IL-1RAP, we modeled transduction of different AML tumor cell lines screened for density of antigenic sites with our lentiviral vectors carrying a third-generation IL-1RAP CAR, an iCASP9 suicide gene, and a truncated CD19 surface gene. We demonstrated that primary AML blasts can be easily transduced (74.55 ± 21.29%, n = 4) and that CAR T cytotoxicity to IL-1RAP is inversely correlated with epitope masking in relation to the number of antigenic sites expressed on the surface of IL-1RAP+ lines. Importantly, we showed that, in vitro, a 24-h exposure of IL-1RAP+/CAR+ leukemia lines to Rimiducid eliminated >85% of the cells. We confirmed that the expression of IL-1RAP CAR by an IL-1RAP+ leukemic cell, by decreasing the membrane availability of the targeted antigen, can induce resistance while a high epitope density maintains sensitivity to CAR T cells. Moreover, the presence of the iCASP9/Rimiducid suicide system safety switch makes this immunotherapy approach safe for application in a future phase 1 clinical trial.
Collapse
Affiliation(s)
- Walid Warda
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Mathieu Neto Da Rocha
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Rim Trad
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Rafik Haderbache
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Yahya Salma
- Laboratory of Applied Biotechnology (LBA3B), AZM Center for Research in Biotechnology and its Applications, Lebanese University, Tripoli, 1300, Lebanon
| | - Lucie Bouquet
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Xavier Roussel
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France.,Hematology Department, Hôpital Jean Minjoz, 25000, Besançon, France
| | - Clémentine Nicod
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Marina Deschamps
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Christophe Ferrand
- INSERM UMR1098 Right, EFS BFC, Univ. Bourgogne Franche-Comté, 25000, Besançon, France.
| |
Collapse
|
571
|
Innamarato P, Pilon-Thomas S. Reactive myelopoiesis and the onset of myeloid-mediated immune suppression: Implications for adoptive cell therapy. Cell Immunol 2020; 361:104277. [PMID: 33476931 DOI: 10.1016/j.cellimm.2020.104277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023]
Abstract
Adoptive T cell therapy (ACT) in combination with lymphodepleting chemotherapy is an effective strategy to induce the eradication of cancer, providing long-term regressions in patients. However, only a minority of patients that receive ACT with tumor-specific T cells exhibit durable benefit. Thus, there is an urgent need to characterize mechanisms of resistance and define strategies to alleviate immunosuppression in the context of ACT in cancer. This article reviews the importance of lymphodepleting regimens in promoting the optimal engraftment and expansion of T cells in hosts after adoptive transfer. In addition, we discuss the role of concomitant immunosuppression and the accumulation of myeloid derived suppressor cells (MDSCs) during immune recovery after lymphodepleting regimens and mobilization regimens.
Collapse
Affiliation(s)
- Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
572
|
Wang K, Zhao Y, Wang X, Wang B, Qin M, Zhu G, Wu H, Liu Z, Zheng X, Zheng H, Chen Z. Case Report: Humanized Selective CD19CAR-T Treatment Induces MRD-Negative Remission in a Pediatric B-ALL Patient With Primary Resistance to Murine-Based CD19CAR-T Therapy. Front Immunol 2020; 11:581116. [PMID: 33424835 PMCID: PMC7786099 DOI: 10.3389/fimmu.2020.581116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/20/2020] [Indexed: 11/27/2022] Open
Abstract
Background CD19 chimeric antigen receptor T cell (CD19CAR-T) has shown great potential to treat acute B cell lymphoblastic leukemia (B-ALL) and B cell lymphoma, and most of anti-CD19 scFv are derived from murine antibody sequences. However, about 10-20% of B-ALL patients exhibit primary resistance to murine-based CD19CAR-T (CD19mCAR-T). Herein, we report that a humanized selective CD19CAR-T (CD19hsCAR-T) may offer a solution to this problem. Case Description A 10-year old boy was diagnosed with high-risk B-ALL in Mar., 2013, and relapsed in Oct., 2018, after he underwent haplo-identical hematopoietic stem cell transplantation (HSCT) in 2017. The patient then received haplo-identical CD19mCAR-T infusions twice following induction chemotherapy with Vincristine, Dexamethasone and Asparaginase (VDL), but no response was observed. We further treated this patient with CD19hsCAR-T following chemotherapy with Vindesine, Idarubicin, Dexamethasone, and Pegylated Asparaginase (VDLD) plus bortezomib. The patient achieved minimal residual disease-negative (MRDneg) complete remission with incomplete hematopoietic recovery (CRi), and remained in CRi for more than 8 months with manageable side effect. The patient, unfortunately, died of unidentified pulmonary infection on Jan. 25 2020. Conclusion CD19hsCAR-T may have the potential to induce remission in patients who are primarily refractory to CD19mCAR-T.
Collapse
Affiliation(s)
- Kai Wang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Xuan Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Bin Wang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Maoquan Qin
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Guanghua Zhu
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Xueling Zheng
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Huyong Zheng
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics (Capital Medical University), Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
573
|
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of adoptive cellular immunotherapy targeting CD19 in its most advanced form. Up to 30% of infused patients achieve long-term survival, meaning that 70% of patients still fail to respond or relapse after therapy. This review will address the unresolved issues relating to responders' characterization, relapse prediction, and prevention, CAR T-cell construct optimization, rational combination with other therapies and treatment toxicity, focusing on the management of relapsed/refractory lymphoma patients. RECENT FINDINGS Many new antigenic targets are currently investigated and raise the hope of broader successes. However, literature data report that treatment failure is not only related to CAR T construct and infusion but is also due to hostile tumor microenvironment and poor interaction with the host effector cells. Further research should not only target CAR T structure, toxicity and associated therapies, but also tumor-related and host-related microenvironment interactions that lead to treatment failure in relapsed/refractory lymphoma patients. SUMMARY Poor persistence of CAR T and loss of CD19 antigen are well established mechanisms of relapse in Acute Lymphoblastic Leukemia (ALL). A fourth generation of CAR T construct is currently investigated to overcome this issue. In non-Hodgkin lymphoma, mechanisms of treatment failure remain poorly understood but tumor and host microenvironment are undoubtedly involved and should be further investigated. A deeper understanding of CAR T-cell therapy failure in individuals will help personalize CAR T-cell therapy in the future.
Collapse
|
574
|
Patriarca A, Gaidano G. Investigational drugs for the treatment of diffuse large B-cell lymphoma. Expert Opin Investig Drugs 2020; 30:25-38. [PMID: 33295827 DOI: 10.1080/13543784.2021.1855140] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Diffuse large B cell lymphoma (DLBCL) is the most frequent lymphoma in adults. 30-40% DLBCL eventually relapse and 10% are primary refractory, posing an unmet clinical need, especially in patients not eligible for hematopoietic stem cell transplant. Knowledge of DLBCL molecular pathogenesis has identified druggable molecular pathways. Surface antigens can be targeted by novel antibodies and innovative cell therapies. Areas covered: This review illuminates those investigational drugs and cell therapies that are currently in early phase clinical trials for the treatment of DLBCL. New small molecules that modulate the pathways involved in the molecular pathogenesis of DLBCL, monospecific and bispecific monoclonal antibodies, drug-immunoconjugates, and cellular therapies are placed under the spotlight. A futuristic perspective concludes the paper. Expert opinion: A precision medicine strategy based on robust molecular predictors of outcome is desirable in the development of investigational small molecules for DLBCL. Novel monoclonal and bispecific antibodies may be offered to (i) relapsed/refractory patients ineligible for CAR-T cells because of comorbidities, and (ii) younger patients before CAR-T cell infusion to reduce a high tumor burden. A focus on the optimal sequencing of the emerging DLBCL drugs is appropriate and necessary.
Collapse
Affiliation(s)
- Andrea Patriarca
- Division of Hematology, Department of Translational Medicine, Università Del Piemonte Orientale and Ospedale Maggiore Della Carità , Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università Del Piemonte Orientale and Ospedale Maggiore Della Carità , Novara, Italy
| |
Collapse
|
575
|
Upadhyay R, Boiarsky JA, Pantsulaia G, Svensson-Arvelund J, Lin MJ, Wroblewska A, Bhalla S, Scholler N, Bot A, Rossi JM, Sadek N, Parekh S, Lagana A, Baccarini A, Merad M, Brown BD, Brody JD. A Critical Role for Fas-Mediated Off-Target Tumor Killing in T-cell Immunotherapy. Cancer Discov 2020; 11:599-613. [PMID: 33334730 DOI: 10.1158/2159-8290.cd-20-0756] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/11/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.
Collapse
Affiliation(s)
- Ranjan Upadhyay
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jonathan A Boiarsky
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gvantsa Pantsulaia
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judit Svensson-Arvelund
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthew J Lin
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aleksandra Wroblewska
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sherry Bhalla
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Norah Sadek
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir Parekh
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessandro Lagana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessia Baccarini
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian D Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua D Brody
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York. .,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
576
|
Filin IY, Solovyeva VV, Kitaeva KV, Rutland CS, Rizvanov AA. Current Trends in Cancer Immunotherapy. Biomedicines 2020; 8:biomedicines8120621. [PMID: 33348704 PMCID: PMC7766207 DOI: 10.3390/biomedicines8120621] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
The search for an effective drug to treat oncological diseases, which have become the main scourge of mankind, has generated a lot of methods for studying this affliction. It has also become a serious challenge for scientists and clinicians who have needed to invent new ways of overcoming the problems encountered during treatments, and have also made important discoveries pertaining to fundamental issues relating to the emergence and development of malignant neoplasms. Understanding the basics of the human immune system interactions with tumor cells has enabled new cancer immunotherapy strategies. The initial successes observed in immunotherapy led to new methods of treating cancer and attracted the attention of the scientific and clinical communities due to the prospects of these methods. Nevertheless, there are still many problems that prevent immunotherapy from calling itself an effective drug in the fight against malignant neoplasms. This review examines the current state of affairs for each immunotherapy method, the effectiveness of the strategies under study, as well as possible ways to overcome the problems that have arisen and increase their therapeutic potentials.
Collapse
Affiliation(s)
- Ivan Y. Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
| | - Catrin S. Rutland
- Faculty of Medicine and Health Science, University of Nottingham, Nottingham NG7 2QL, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.Y.F.); (V.V.S.); (K.V.K.)
- Republic Clinical Hospital, 420064 Kazan, Russia
- Correspondence: ; Tel.: +7-905-316-7599
| |
Collapse
|
577
|
Yang M, Wang L, Ni M, Neuber B, Wang S, Gong W, Sauer T, Sellner L, Schubert ML, Hückelhoven-Krauss A, Hong J, Zhu L, Kleist C, Eckstein V, Müller-Tidow C, Dreger P, Schmitt M, Schmitt A. Pre-sensitization of Malignant B Cells Through Venetoclax Significantly Improves the Cytotoxic Efficacy of CD19.CAR-T Cells. Front Immunol 2020; 11:608167. [PMID: 33362794 PMCID: PMC7756123 DOI: 10.3389/fimmu.2020.608167] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promising responses in patients with refractory or relapsed aggressive B-cell malignancies that are resistant to conventional chemotherapy or stem cell transplantation. A potentially combinatorial therapeutic strategy may be the inhibition of anti-apoptotic Bcl-2 family proteins, overexpressed in most cancer cells. In this study we investigated the combination of 3rd-generation CD19.CAR-T cells and the BH3 mimetics venetoclax, a Bcl-2 inhibitor, or S63845, a Mcl-1 inhibitor, under three different treatment conditions: pre-sensitization of cancer cells with BH3 mimetics followed by CAR-T cell treatment, simultaneous combination therapy, and the administration of BH3 mimetics after CAR-T cell treatment. Our results showed that administration of CAR-T cells and BH3 mimetics had a significant effect on the quantity and quality of CD19.CAR-T cells. The administration of BH3 mimetics prior to CAR-T cell therapy exerted an enhanced cytotoxic efficacy by upregulating the CD19 expression and pro-apoptotic proteins in highly sensitive tumor cells, and thereby improving both CD19.CAR-T cell cytotoxicity and persistence. In simultaneous and post-treatment approaches, however, the quantity of CAR-T cells was adversely affected. Our findings indicate pre-sensitization of highly sensitive tumor cells with BH3 mimetics could enhance the cytotoxic efficacy of CAR-T cell treatment.
Collapse
Affiliation(s)
- Mingya Yang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Sanmei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Wenjie Gong
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tim Sauer
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,Oncology Business Unit-Medical Affairs, Takeda Pharma Vertrieb GmbH & Co. KG, Berlin, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Angela Hückelhoven-Krauss
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jian Hong
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lixin Zhu
- Department of General Surgery and Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Christian Kleist
- Department of Nuclear Medicine, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
578
|
Gaissmaier L, Christopoulos P. Immune Modulation in Lung Cancer: Current Concepts and Future Strategies. Respiration 2020; 99:1-27. [PMID: 33291116 DOI: 10.1159/000510385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy represents the most dynamic field of biomedical research currently, with thoracic immuno-oncology as a forerunner. PD-(L)1 inhibitors are already part of standard first-line treatment for both non-small-cell and small-cell lung cancer, while unprecedented 5-year survival rates of 15-25% have been achieved in pretreated patients with metastatic disease. Evolving strategies are mainly aiming for improvement of T-cell function, increase of immune activation in the tumor microenvironment (TME), and supply of tumor-reactive lymphocytes. Several novel therapeutics have demonstrated preclinical efficacy and are increasingly used in rational combinations within clinical trials. Two overarching trends dominate: extension of immunotherapy to earlier disease stages, mainly as neoadjuvant treatment, and a shift of focus towards multivalent, individualized, mutatome-based antigen-specific modalities, mainly adoptive cell therapies and cancer vaccines. The former ensures ample availability of treated and untreated patient samples, the latter facilitates deeper mechanistic insights, and both in combination build an overwhelming force that is accelerating progress and driving the greatest revolution cancer medicine has seen so far. Today, immune modulation represents the most potent therapeutic modality in oncology, the most important topic in clinical and translational cancer research, and arguably our greatest, meanwhile justified hope for achieving cure of pulmonary neoplasms and other malignancies in the next future.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany,
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany,
| |
Collapse
|
579
|
Basar R, Daher M, Rezvani K. Next-generation cell therapies: the emerging role of CAR-NK cells. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:570-578. [PMID: 33275752 PMCID: PMC7727537 DOI: 10.1182/hematology.2020002547] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
T cells engineered with chimeric antigen receptors (CARs) have revolutionized the field of cell therapy and changed the paradigm of treatment for many patients with relapsed or refractory B-cell malignancies. Despite this progress, there are limitations to CAR-T cell therapy in both the autologous and allogeneic settings, including practical, logistical, and toxicity issues. Given these concerns, there is a rapidly growing interest in natural killer cells as alternative vehicles for CAR engineering, given their unique biological features and their established safety profile in the allogeneic setting. Other immune effector cells, such as invariant natural killer T cells, γδ T cells, and macrophages, are attracting interest as well and eventually may be added to the repertoire of engineered cell therapies against cancer. The pace of these developments will undoubtedly benefit from multiple innovative technologies, such as the CRISPR-Cas gene editing system, which offers great potential to enhance the natural ability of immune effector cells to eliminate refractory cancers.
Collapse
Affiliation(s)
- Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
580
|
Acharya UH, Walter RB. Chimeric Antigen Receptor (CAR)-Modified Immune Effector Cell Therapy for Acute Myeloid Leukemia (AML). Cancers (Basel) 2020; 12:E3617. [PMID: 33287224 PMCID: PMC7761730 DOI: 10.3390/cancers12123617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Despite the availability of an increasing number of targeted therapeutics and wider use of allogeneic hematopoietic stem cell transplantation, many patients with acute myeloid leukemia (AML) ultimately succumb to this disease. Given their remarkable efficacy in B-acute lymphoblastic leukemia and other CD19-expressing B cell malignancies, there is hope adoptive cellular transfer, particularly chimeric antigen receptor (CAR)-modified immune effector cell (IEC) therapies, may afford a novel, potent immune-based approach for the treatment of AML that complements or replaces existing ones and improves cure rates. However, it is unclear how best to translate the success of these therapies from B cell malignancies, where use of highly potent immunotherapies is facilitated by identified target antigens with near ubiquitous expression on malignant cells and non-fatal consequences from "on-target, off-tumor cell" toxicities. Herein, we review the current status of CAR-modified IEC therapies for AML, with considerations regarding suitable, relatively leukemia-restricted target antigens, expected toxicities, and interactions of the engineered cells with a profoundly immunosuppressive tumor microenvironment that restricts their therapeutic efficacy. With these challenges in mind, we will discuss possible strategies to improve the cells' potency as well as their therapeutic window for optimal clinical use in AML.
Collapse
Affiliation(s)
- Utkarsh H. Acharya
- Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
581
|
Feucht J, Sadelain M. Function and evolution of the prototypic CD28ζ and 4-1BBζ chimeric antigen receptors. ACTA ACUST UNITED AC 2020; 8:2-11. [PMID: 35757562 PMCID: PMC9216534 DOI: 10.1016/j.iotech.2020.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) specific for CD19 have yielded remarkable clinical outcomes in patients with refractory B-cell malignancies. The first CARs to be approved by the US Food and Drug Administration and the European Medicines Agency are CD19 CARs that comprise either CD28/CD3ζ or 4-1BB/CD3ζ dual-signalling domains. While their efficacy and safety profiles in patients with B-cell malignancies are comparable overall, the functional properties these two CAR designs impart upon engineered T cells differ significantly. Remarkably, alternative costimulatory domains have not, to date, superseded these foundational designs. Rather, recent CAR advances have focused on perfecting the original CD28- and 4-1BB-based CD19 CARs by calibrating strength of activation, pre-empting T-cell exhaustion and increasing the functional persistence of CAR T cells. This article reviews the essential biological properties of these first-in-class prototypes and their recent evolution. CD19 chimeric antigen receptor (CAR) therapy has shown remarkable success against B-cell malignancies. The prototypic CD19 CARs comprise either CD28/CD3ζ or 4-1BB/CD3ζ signalling domains. Both CD19 CARs yield similar efficacy but impart distinct T-cell functionalities. Novel CAR designs aim to enhance the persistence or effector potency of T cells. Genome editing averts variegated CAR expression and sustains T-cell function.
Collapse
Affiliation(s)
| | - M. Sadelain
- Correspondence to: Michel Sadelain, Director, Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. Tel: 212-639-6190
| |
Collapse
|
582
|
Abstract
Therapeutic resistance continues to be an indominable foe in our ambition for curative cancer treatment. Recent insights into the molecular determinants of acquired treatment resistance in the clinical and experimental setting have challenged the widely held view of sequential genetic evolution as the primary cause of resistance and brought into sharp focus a range of non-genetic adaptive mechanisms. Notably, the genetic landscape of the tumour and the non-genetic mechanisms used to escape therapy are frequently linked. Remarkably, whereas some oncogenic mutations allow the cancer cells to rapidly adapt their transcriptional and/or metabolic programme to meet and survive the therapeutic pressure, other oncogenic drivers convey an inherent cellular plasticity to the cancer cell enabling lineage switching and/or the evasion of anticancer immunosurveillance. The prevalence and diverse array of non-genetic resistance mechanisms pose a new challenge to the field that requires innovative strategies to monitor and counteract these adaptive processes. In this Perspective we discuss the key principles of non-genetic therapy resistance in cancer. We provide a perspective on the emerging data from clinical studies and sophisticated cancer models that have studied various non-genetic resistance pathways and highlight promising therapeutic avenues that may be used to negate and/or counteract the non-genetic adaptive pathways.
Collapse
Affiliation(s)
- Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium.
- Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Center for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia.
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Center for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
583
|
Li J, Luo Y, Li B, Xia Y, Wang H, Fu C. Implantable and Injectable Biomaterial Scaffolds for Cancer Immunotherapy. Front Bioeng Biotechnol 2020; 8:612950. [PMID: 33330440 PMCID: PMC7734317 DOI: 10.3389/fbioe.2020.612950] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy has become an emerging strategy recently producing durable immune responses in patients with varieties of malignant tumors. However, the main limitation for the broad application of immunotherapies still to reduce side effects by controlling and regulating the immune system. In order to improve both efficacy and safety, biomaterials have been applied to immunotherapies for the specific modulation of immune cells and the immunosuppressive tumor microenvironment. Recently, researchers have constantly developed biomaterials with new structures, properties and functions. This review provides the most recent advances in the delivery strategies of immunotherapies based on localized biomaterials, focusing on the implantable and injectable biomaterial scaffolds. Finally, the challenges and prospects of applying implantable and injectable biomaterial scaffolds in the development of future cancer immunotherapies are discussed.
Collapse
Affiliation(s)
- Jie Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yiqian Luo
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Baoqin Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
584
|
Parayath NN, Stephan SB, Koehne AL, Nelson PS, Stephan MT. In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo. Nat Commun 2020; 11:6080. [PMID: 33247092 PMCID: PMC7695830 DOI: 10.1038/s41467-020-19486-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Engineering chimeric antigen receptors (CAR) or T cell receptors (TCR) helps create disease-specific T cells for targeted therapy, but the cost and rigor associated with manufacturing engineered T cells ex vivo can be prohibitive, so programing T cells in vivo may be a viable alternative. Here we report an injectable nanocarrier that delivers in vitro-transcribed (IVT) CAR or TCR mRNA for transiently reprograming of circulating T cells to recognize disease-relevant antigens. In mouse models of human leukemia, prostate cancer and hepatitis B-induced hepatocellular carcinoma, repeated infusions of these polymer nanocarriers induce sufficient host T cells expressing tumor-specific CARs or virus-specific TCRs to cause disease regression at levels similar to bolus infusions of ex vivo engineered lymphocytes. Given their ease of manufacturing, distribution and administration, these nanocarriers, and the associated platforms, could become a therapeutic for a wide range of diseases. Ex vivo engineering of antigen-specific T cells has shown therapeutic efficacy but can be costly and scarce. Here the authors show that in vitro-transcribed antigen receptor mRNA packaged in nanocarriers can directly induce, in vivo, transient their expression in circulating T cells to provide therapeutic effects in mouse models of cancer or viral infection.
Collapse
Affiliation(s)
- N N Parayath
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - S B Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - A L Koehne
- Translational Pathology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - P S Nelson
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - M T Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA. .,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, 98195, USA. .,Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
585
|
Zhu X, Li S, Xu B, Luo H. Cancer evolution: A means by which tumors evade treatment. Biomed Pharmacother 2020; 133:111016. [PMID: 33246226 DOI: 10.1016/j.biopha.2020.111016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
Although various methods have been tried to study and treat cancer, the cancer remains a major challenge for human medicine today. One important reason for this is the presence of cancer evolution. Cancer evolution is a process in which tumor cells adapt to the external environment, which can suppress the human immune system's ability to recognize and attack tumors, and also reduce the reproducibility of cancer research. Among them, heterogeneity of the tumor provides intrinsic motivation for this process. Recently, with the development of related technologies such as liquid biopsy, more and more knowledge about cancer evolution has been gained and interest in this topic has also increased. Therefore, starting from the causes of tumorigenesis, this paper introduces several tumorigenesis processes and pathways, as well as treatment options for different targets.
Collapse
Affiliation(s)
- Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Shi Li
- Guangdong Key Laboratory of Urogenital Tumor Systems and Synthetic Biology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China; Shenzhen Key Laboratory of Genitourinary Tumor, Translational Medicine Institute of Shenzhen, The Second People's Hospital of Shenzhen, Shenzhen, China; College of Bioengineering, Chongqing University, Chongqing, China
| | - Bairui Xu
- The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China.
| |
Collapse
|
586
|
Ghilardi G, Braendstrup P, Chong EA, Schuster SJ, Svoboda J, Ruella M. CAR-T TREK through the lymphoma universe, to boldly go where no other therapy has gone before. Br J Haematol 2020; 193:449-465. [PMID: 33222167 DOI: 10.1111/bjh.17191] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T cells (CART) therapies have changed and continue to change the treatment paradigms for B-cell malignancies because they can achieve durable complete remission in patients in whom multiple lines of treatment have failed. These unprecedented results have led to the widespread use of anti-CD19 CART therapy for patients with relapsed and refractory aggressive large B-cell lymphomas. While long-term follow-up data show that about one-third of patients achieve prolonged complete remission and are potentially cured, the majority of patients either do not respond to CD19 CART therapy or eventually relapse after CD19 CART therapy. These results are, on the one hand, driving intense research into identifying mechanisms of relapse and, on the other hand, inspiring the development of novel strategies to overcome resistance. This review summarizes current clinical outcomes of CART immunotherapy in B-cell non-Hodgkin lymphomas, describes the most up-to-date understanding of mechanisms of relapse and discusses novel strategies to address resistance to CART therapy. We are indeed at the beginning of a scientific trek to explore the mechanisms of resistance, seek out new, more effective treatment approaches based on these discoveries and to boldly go where no other therapy has gone before!
Collapse
Affiliation(s)
- Guido Ghilardi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Braendstrup
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Elise A Chong
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
587
|
McComb S, Lee SH. Current Advances and Hurdles in Chimeric Antigen Receptor Technology. Cancers (Basel) 2020; 12:cancers12113329. [PMID: 33187171 PMCID: PMC7697884 DOI: 10.3390/cancers12113329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/07/2020] [Indexed: 11/16/2022] Open
Abstract
Since tumor-specific T cells were first utilized to treat melanoma patients in 1986 [...].
Collapse
Affiliation(s)
- Scott McComb
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON K1H 8M5, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
588
|
Yang Y, Li Y, Gu H, Dong M, Cai Z. Emerging agents and regimens for multiple myeloma. J Hematol Oncol 2020; 13:150. [PMID: 33168044 PMCID: PMC7654052 DOI: 10.1186/s13045-020-00980-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
The outcomes of multiple myeloma (MM) have been improved significantly with the therapies incorporating proteasome inhibitors (PI), immunomodulatory drugs, monoclonal antibodies (MoAb) and stem cell transplantation. However, relapsed and refractory MM (RRMM) remains a major challenge. Novel agents and regimens are under active clinical development. These include new PIs such as ixazomib, marizomib, and oprozomib; new MoAbs such as isatuximab and MOR202; novel epigenetic agent ricolinostat and novel cytokines such as siltuximab. Recently, the first XPO-1 inhibitor, selinexor, was approved for RRMM. BCMA-targeted BiTE, antibody-drug conjugates and CAR-T cells have the potential to revolutionize the therapy for RRMM. In this review, we summarized the latest clinical development of these novel agents and regimens.
Collapse
Affiliation(s)
- Yang Yang
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Li
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengmeng Dong
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
589
|
Liu Y, Zhang T, Zhang H, Li J, Zhou N, Fiskesund R, Chen J, Lv J, Ma J, Zhang H, Tang K, Cheng F, Zhou Y, Zhang X, Wang N, Huang B. Cell Softness Prevents Cytolytic T-cell Killing of Tumor-Repopulating Cells. Cancer Res 2020; 81:476-488. [PMID: 33168645 DOI: 10.1158/0008-5472.can-20-2569] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/23/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
Biomechanics is a fundamental feature of a cell. However, the manner by which actomysin tension affects tumor immune evasion remains unclear. Here we show that although cytotoxic T lymphocytes (CTL) can effectively destroy stiff differentiated tumor cells, they fail to kill soft tumor-repopulating cells (TRC). TRC softness prevented membrane pore formation caused by CTL-released perforin. Perforin interacting with nonmuscle myosin heavy-chain 9 transmitted forces to less F-actins in soft TRC, thus generating an inadequate contractile force for perforin pore formation. Stiffening TRC allowed perforin the ability to drill through the membrane, leading to CTL-mediated killing of TRC. Importantly, overcoming mechanical softness in human TRC also enhanced TRC cell death caused by human CTL, potentiating a mechanics-based immunotherapeutic strategy. These findings reveal a mechanics-mediated tumor immune evasion, thus potentially providing an alternative approach for tumor immunotherapy. SIGNIFICANCE: Tumor-repopulating cells evade CD8+ cytolytic T-cell killing through a mechanical softness mechanism, underlying the impediment of perforin pore formation at the immune synapse site.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China.
- Clinical Immunology Center, CAMS, Beijing, China
| | - Tianzhen Zhang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Haizeng Zhang
- National Cancer Center/Cancer Hospital, CAMS, Beijing, China
| | - Jiping Li
- Beijing Smartchip Microelectronics Technology Company Limited, Beijing, China
| | - Nannan Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Roland Fiskesund
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
- Karolinska Institutet Medical School, Stockholm, Sweden
| | - Junwei Chen
- Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiadi Lv
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Jingwei Ma
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huafeng Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feiran Cheng
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Yabo Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Ning Wang
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Bo Huang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China.
- Clinical Immunology Center, CAMS, Beijing, China
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
590
|
Grünewald TGP, Alonso M, Avnet S, Banito A, Burdach S, Cidre‐Aranaz F, Di Pompo G, Distel M, Dorado‐Garcia H, Garcia‐Castro J, González‐González L, Grigoriadis AE, Kasan M, Koelsche C, Krumbholz M, Lecanda F, Lemma S, Longo DL, Madrigal‐Esquivel C, Morales‐Molina Á, Musa J, Ohmura S, Ory B, Pereira‐Silva M, Perut F, Rodriguez R, Seeling C, Al Shaaili N, Shaabani S, Shiavone K, Sinha S, Tomazou EM, Trautmann M, Vela M, Versleijen‐Jonkers YMH, Visgauss J, Zalacain M, Schober SJ, Lissat A, English WR, Baldini N, Heymann D. Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020; 12:e11131. [PMID: 33047515 PMCID: PMC7645378 DOI: 10.15252/emmm.201911131] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are heterogeneous and clinically challenging soft tissue and bone cancers. Although constituting only 1% of all human malignancies, sarcomas represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. More than 100 histological subtypes have been characterized to date, and many more are being discovered due to molecular profiling. Owing to their mostly aggressive biological behavior, relative rarity, and occurrence at virtually every anatomical site, many sarcoma subtypes are in particular difficult-to-treat categories. Current multimodal treatment concepts combine surgery, polychemotherapy (with/without local hyperthermia), irradiation, immunotherapy, and/or targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the latest advances in the molecular biology of sarcomas and their effects on clinical oncology; it is meant for a broad readership ranging from novices to experts in the field of sarcoma.
Collapse
Affiliation(s)
- Thomas GP Grünewald
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Division of Translational Pediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), Hopp Children's Cancer Center (KiTZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Marta Alonso
- Program in Solid Tumors and BiomarkersFoundation for the Applied Medical ResearchUniversity of Navarra PamplonaPamplonaSpain
| | - Sofia Avnet
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Ana Banito
- Pediatric Soft Tissue Sarcoma Research GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Stefan Burdach
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Florencia Cidre‐Aranaz
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | - Gemma Di Pompo
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | | | | | | | | | - Merve Kasan
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | | | - Fernando Lecanda
- Division of OncologyAdhesion and Metastasis LaboratoryCenter for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Silvia Lemma
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Dario L Longo
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | | | | | - Julian Musa
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Shunya Ohmura
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | - Miguel Pereira‐Silva
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Francesca Perut
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Rene Rodriguez
- Instituto de Investigación Sanitaria del Principado de AsturiasOviedoSpain
- CIBER en oncología (CIBERONC)MadridSpain
| | | | - Nada Al Shaaili
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Shabnam Shaabani
- Department of Drug DesignUniversity of GroningenGroningenThe Netherlands
| | - Kristina Shiavone
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Snehadri Sinha
- Department of Oral and Maxillofacial DiseasesUniversity of HelsinkiHelsinkiFinland
| | | | - Marcel Trautmann
- Division of Translational PathologyGerhard‐Domagk‐Institute of PathologyMünster University HospitalMünsterGermany
| | - Maria Vela
- Hospital La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | | | | | - Marta Zalacain
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | - Sebastian J Schober
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Andrej Lissat
- University Children′s Hospital Zurich – Eleonoren FoundationKanton ZürichZürichSwitzerland
| | - William R English
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Nicola Baldini
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Dominique Heymann
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Université de NantesInstitut de Cancérologie de l'OuestTumor Heterogeneity and Precision MedicineSaint‐HerblainFrance
| |
Collapse
|
591
|
Rodríguez-Otero P, Prósper F, Alfonso A, Paiva B, Miguel JFS. CAR T-Cells in Multiple Myeloma Are Ready for Prime Time. J Clin Med 2020; 9:E3577. [PMID: 33172026 PMCID: PMC7694626 DOI: 10.3390/jcm9113577] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 01/22/2023] Open
Abstract
The survival of patients with multiple myeloma (MM) has been dramatically improved in the last decade thanks to the incorporation of second-generation proteasome inhibitors (PI), immunomodulatory drugs (IMID), and, more recently, anti-CD38 monoclonal antibodies (MoAb). Nevertheless, still, a major proportion of MM patients will relapse, underscoring the need for new therapies in this disease. Moreover, survival in patients failing the current standard of care regimens (including PI, IMIDs, and anti-CD38 MoAb), which is now defined as triple-class refractory, remains dismal, and new drugs with different mechanism of action are needed. B-cell maturation antigen (BCMA)-targeted therapies and in particular chimeric antigen receptor T cell (CAR T-cell) treatment have emerged as promising platforms to overcome refractoriness to conventional drugs. In this manuscript, we review the current available data regarding CAR T-cell therapy for MM, with a special focus on target selection, clinical results, limitations, and future strategies.
Collapse
Affiliation(s)
- Paula Rodríguez-Otero
- Clínica Universidad de Navarra, Centro de investigación médica aplicada (Cima), CIBERONC, IDISNA, 31008 Pamplona, Spain; (F.P.); (A.A.); (B.P.); (J.F.S.M.)
| | | | | | | | | |
Collapse
|
592
|
Prinzing B, Schreiner P, Bell M, Fan Y, Krenciute G, Gottschalk S. MyD88/CD40 signaling retains CAR T cells in a less differentiated state. JCI Insight 2020; 5:136093. [PMID: 33148882 PMCID: PMC7710311 DOI: 10.1172/jci.insight.136093] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy for solid tumors has shown limited efficacy in early-phase clinical studies. The majority of CARs encode CD28 and/or 41BB costimulatory endodomains, and we explored whether MyD88 and CD40 (MC) costimulatory endodomains in CARs could improve their antitumor activity. We generated CD28-, 41BB-, and MC-CAR T cells and demonstrated that MC-CAR T cells have greater proliferative capacity and antitumor activity in repeat stimulation assays and in tumor models in vivo. Transcriptomic analysis revealed that MC-CAR T cells expressed higher levels of MYB and FOXM1, key cell cycle regulators, and were activated at baseline. After stimulation, MC-CAR T cells remained in a less differentiated state than CD28- and 41BB-CAR T cells as judged by low levels of transcription factor TBET and B lymphocyte induced maturation protein 1 expression and lower cytolytic activity in comparison with CD28- and 41BB-CAR T cells. Thus, including MyD88 and CD40 signaling domains in CARs may improve current CAR T cell therapy approaches for solid tumors.
Collapse
Affiliation(s)
- Brooke Prinzing
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | | | - Matthew Bell
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
593
|
Xie Y, Hu Y, Zhou N, Yao C, Wu L, Liu L, Chen F. CAR T-cell therapy for triple-negative breast cancer: Where we are. Cancer Lett 2020; 491:121-131. [PMID: 32795486 DOI: 10.1016/j.canlet.2020.07.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most complex and challenging breast cancer subtype to treat, and chemotherapy remains the standard of care. Clinically, TNBC has a relatively high rate of recurrence and poor prognosis, which leads to a significant effort to discover novel strategies to treat patients with these tumors. Currently, chimeric antigen receptor (CAR) T cell-based immunotherapy redirects the patient's immune system directly to recognize and eradicate tumor-associated antigens (TAAs) expressing tumor cells being explored as a treatment for TNBC. A steadily increasing research in CAR T-cell therapy targeting different TAAs in TNBC has reported. In this review, we introduce the CAR technology and summarize the potential TAAs, available CARs, the antitumor activity, and the related toxicity of CARs currently under investigation for TNBC. We also highlight the potential strategies to prevent/reduce potential "on target, off tumor" toxicity induced by CAR T-cell therapy. This review will help to explore proper targets to expand further the CAR T-cell therapy for TNBCs in the clinic.
Collapse
Affiliation(s)
- Yuetao Xie
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Yi Hu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Nawu Zhou
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Cuicui Yao
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Lixin Wu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Lin Liu
- Everest Medical Care, 2010 West Chester Pike, Havertown, PA, 19083, USA
| | - Fang Chen
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China.
| |
Collapse
|
594
|
Voutsadakis IA. Further Understanding of High-Grade Serous Ovarian Carcinogenesis: Potential Therapeutic Targets. Cancer Manag Res 2020; 12:10423-10437. [PMID: 33116896 PMCID: PMC7585777 DOI: 10.2147/cmar.s249540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/25/2020] [Indexed: 01/09/2023] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most common type of ovarian cancer and the most lethal gynecologic malignancy due to advanced stage at presentation. Recent years have witnessed progress in the therapy of HGSOC with the introduction of PARP (poly-adenosine diphosphate ribose polymerase) inhibitors and the anti-angiogenic monoclonal antibody bevacizumab to the backbone of chemotherapy or as maintenance therapy after chemotherapy. The improved molecular understanding of ovarian cancer pathogenesis, which has brought these therapies into the clinic, aspires to extend the boundaries of therapies through elucidation of other molecular aspects of ovarian carcinogenesis. This accumulating knowledge has started to be translated to additional targeted therapies that are in various stages of development. These include inhibitors of the function of other proteins involved in homologous recombination deficiency (HRD), such as WEE1 kinase, ATM/ATR kinases and CDK12 inhibitors. Despite disappointing results with immune checkpoint inhibitors monotherapy, harnessing the immune system in HGSOC with combination therapies that promote antigen production and immune cell activation is an avenue being explored. This paper examines arising HGSOC therapies based on molecular understanding of pathogenesis.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, Ontario, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| |
Collapse
|
595
|
Cai Q, Zhang M, Li Z. Potential strategies against resistance to CAR T-cell therapy in haematological malignancies. Ther Adv Med Oncol 2020; 12:1758835920962963. [PMID: 33133242 PMCID: PMC7576929 DOI: 10.1177/1758835920962963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/07/2020] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a rapidly developing method for adoptive immunotherapy of tumours in recent years. CAR T-cell therapies have demonstrated unprecedented efficacy in the treatment of patients with haematological malignancies. A 90% complete response (CR) rate has been reported in patients with advanced relapse or refractory acute lymphoblastic leukaemia, while >50% CR rates have been reported in cases of chronic lymphocytic leukaemia and partial B-cell lymphoma. Despite the high CR rates, a subset of the patients with complete remission still relapse. The mechanism of development of resistance is not clearly understood. Some patients have been reported to demonstrate antigen-positive relapse, whereas others show antigen-negative relapses. Patients who relapse following CAR T-cell therapy, have very poor prognosis and novel approaches to overcome resistance are required urgently. Herein, we have reviewed current literature and research that have investigated the strategies to overcome resistance to CAR T-cell therapy.
Collapse
Affiliation(s)
- Qing Cai
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 6th Floor, Building 10, No.1 Construction East Road, Zhengzhou, Henan Province 450052, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
596
|
Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat Med 2020; 26:1569-1575. [PMID: 33020647 DOI: 10.1038/s41591-020-1081-3] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/26/2020] [Indexed: 01/09/2023]
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 are a breakthrough treatment for relapsed, refractory B cell malignancies1-5. Despite impressive outcomes, relapse with CD19- disease remains a challenge. We address this limitation through a first-in-human trial of bispecific anti-CD20, anti-CD19 (LV20.19) CAR T cells for relapsed, refractory B cell malignancies. Adult patients with B cell non-Hodgkin lymphoma or chronic lymphocytic leukemia were treated on a phase 1 dose escalation and expansion trial (NCT03019055) to evaluate the safety of 4-1BB-CD3ζ LV20.19 CAR T cells and the feasibility of on-site manufacturing using the CliniMACS Prodigy system. CAR T cell doses ranged from 2.5 × 105-2.5 × 106 cells per kg. Cell manufacturing was set at 14 d with the goal of infusing non-cryopreserved LV20.19 CAR T cells. The target dose of LV20.19 CAR T cells was met in all CAR-naive patients, and 22 patients received LV20.19 CAR T cells on protocol. In the absence of dose-limiting toxicity, a dose of 2.5 × 106 cells per kg was chosen for expansion. Grade 3-4 cytokine release syndrome occurred in one (5%) patient, and grade 3-4 neurotoxicity occurred in three (14%) patients. Eighteen (82%) patients achieved an overall response at day 28, 14 (64%) had a complete response, and 4 (18%) had a partial response. The overall response rate to the dose of 2.5 × 106 cells per kg with non-cryopreserved infusion (n = 12) was 100% (complete response, 92%; partial response, 8%). Notably, loss of the CD19 antigen was not seen in patients who relapsed or experienced treatment failure. In conclusion, on-site manufacturing and infusion of non-cryopreserved LV20.19 CAR T cells were feasible and therapeutically safe, showing low toxicity and high efficacy. Bispecific CARs may improve clinical responses by mitigating target antigen downregulation as a mechanism of relapse.
Collapse
|
597
|
Jasinski S, De Los Reyes FA, Yametti GC, Pierro J, Raetz E, Carroll WL. Immunotherapy in Pediatric B-Cell Acute Lymphoblastic Leukemia: Advances and Ongoing Challenges. Paediatr Drugs 2020; 22:485-499. [PMID: 32860590 PMCID: PMC7537790 DOI: 10.1007/s40272-020-00413-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.
Collapse
Affiliation(s)
- Sylwia Jasinski
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | | | - Gloria Contreras Yametti
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Joanna Pierro
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - William L Carroll
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
598
|
Bouziana S, Bouzianas D. Anti-CD19 CAR-T cells: Digging in the dark side of the golden therapy. Crit Rev Oncol Hematol 2020; 157:103096. [PMID: 33181441 DOI: 10.1016/j.critrevonc.2020.103096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/25/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
The unprecedented technological advances in genetic engineering have resulted in the advent of the very promising chimeric antigen receptor (CAR)-T cell therapy. Based on the striking outcomes of clinical trials, the first two commercial CAR-T cell products, tisagenlecleucel and axicabtagene ciloleucel, have been approved in both the United States and Europe for the treatment of patients with highly aggressive CD19-positive hematological malignancies. Despite the initial remarkable responses many patients finally relapse, implying the presence of resistance mechanisms. In this review, we describe the limitations and resistance mechanisms to anti-CD19 CAR-T cells and address potential strategies to overcome CAR-T cell barriers.
Collapse
Affiliation(s)
- Stella Bouziana
- Department of Hematology-BMT Unit, G. Papanikolaou Hospital, Thessaloniki, Greece.
| | - Dimitrios Bouzianas
- BReMeL Biopharmaceutical and Regenerative Medicine Laboratories, Thessaloniki, Greece
| |
Collapse
|
599
|
Mane MM, Cohen IJ, Ackerstaff E, Shalaby K, Ijoma JN, Ko M, Maeda M, Albeg AS, Vemuri K, Satagopan J, Moroz A, Zurita J, Shenker L, Shindo M, Nickles T, Nikolov E, Moroz MA, Koutcher JA, Serganova I, Ponomarev V, Blasberg RG. Lactate Dehydrogenase A Depletion Alters MyC-CaP Tumor Metabolism, Microenvironment, and CAR T Cell Therapy. Mol Ther Oncolytics 2020; 18:382-395. [PMID: 32913888 PMCID: PMC7452096 DOI: 10.1016/j.omto.2020.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022] Open
Abstract
To enhance human prostate-specific membrane antigen (hPSMA)-specific chimeric antigen receptor (CAR) T cell therapy in a hPSMA+ MyC-CaP tumor model, we studied and imaged the effect of lactate dehydrogenase A (LDH-A) depletion on the tumor microenvironment (TME) and tumor progression. Effective LDH-A short hairpin RNA (shRNA) knockdown (KD) was achieved in MyC-CaP:hPSMA+ Renilla luciferase (RLuc)-internal ribosome entry site (IRES)-GFP tumor cells, and changes in tumor cell metabolism and in the TME were monitored. LDH-A downregulation significantly inhibited cell proliferation and subcutaneous tumor growth compared to control cells and tumors. However, total tumor lactate concentration did not differ significantly between LDH-A knockdown and control tumors, reflecting the lower vascularity, blood flow, and clearance of lactate from LDH-A knockdown tumors. Comparing treatment responses of MyC-CaP tumors with LDH-A depletion and/or anti-hPSMA CAR T cells showed that the dominant effect on tumor growth was LDH-A depletion. With anti-hPSMA CAR T cell treatment, tumor growth was significantly slower when combined with tumor LDH-A depletion and compared to control tumor growth (p < 0.0001). The lack of a complete tumor response in our animal model can be explained in part by (1) the lower activity of human CAR T cells against hPSMA-expressing murine tumors in a murine host, and (2) a loss of hPSMA antigen from the tumor cell surface in progressive generations of tumor cells.
Collapse
Affiliation(s)
- Mayuresh M. Mane
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ivan J. Cohen
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Khalid Shalaby
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jenny N. Ijoma
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Myat Ko
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Masatomo Maeda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Avi S. Albeg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kiranmayi Vemuri
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jaya Satagopan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Moroz
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Skolkovo Institute of Science and Technology, 143026 Moscow, Russia
| | - Juan Zurita
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Larissa Shenker
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Masahiro Shindo
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tanner Nickles
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ekaterina Nikolov
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxim A. Moroz
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason A. Koutcher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Inna Serganova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vladimir Ponomarev
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ronald G. Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
600
|
Rafia C, Harly C, Scotet E. Beyond CAR T cells: Engineered Vγ9Vδ2 T cells to fight solid tumors. Immunol Rev 2020; 298:117-133. [DOI: 10.1111/imr.12920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Chirine Rafia
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
- ImCheck Therapeutics Marseille France
| | - Christelle Harly
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
| | - Emmanuel Scotet
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
| |
Collapse
|